US8349832B2 - Compounds and compositions for treating cancer - Google Patents

Compounds and compositions for treating cancer Download PDF

Info

Publication number
US8349832B2
US8349832B2 US12/874,306 US87430610A US8349832B2 US 8349832 B2 US8349832 B2 US 8349832B2 US 87430610 A US87430610 A US 87430610A US 8349832 B2 US8349832 B2 US 8349832B2
Authority
US
United States
Prior art keywords
cancer
cell
cells
compounds
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US12/874,306
Other versions
US20110053938A1 (en
Inventor
Michael Andrew Foley
Robert Gould
Peter Elliott
Anna Mandinova
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
General Hospital Corp
Original Assignee
General Hospital Corp
Canthera Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US12/874,306 priority Critical patent/US8349832B2/en
Application filed by General Hospital Corp, Canthera Therapeutics Inc filed Critical General Hospital Corp
Priority to PCT/US2011/025022 priority patent/WO2012030408A1/en
Priority to US13/028,571 priority patent/US8318737B2/en
Publication of US20110053938A1 publication Critical patent/US20110053938A1/en
Priority to US13/162,632 priority patent/US20120157455A1/en
Assigned to CANTHERA THERAPEUTICS, INC. reassignment CANTHERA THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOLEY, MICHAEL A, LEE, SAM, MANDINOVA, ANNA, ELLIOTT, PETER, GOULD, ROBERT
Assigned to MASSACHUSETTS GENERAL HOSPITAL CORPORATION reassignment MASSACHUSETTS GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MANDINOVA, ANNA
Assigned to MANDINOVA, ANNA reassignment MANDINOVA, ANNA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CANTHERA THERAPEUTICS INC.
Assigned to LEE, SAM reassignment LEE, SAM ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CANTHERA THERAPEUTICS INC.
Assigned to MASSACHUSETTS GENERAL HOSPITAL CORPORATION reassignment MASSACHUSETTS GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, SAM
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEE, SAM W.
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MANDINOVA, ANNA
Priority to US13/679,522 priority patent/US20130203757A1/en
Publication of US8349832B2 publication Critical patent/US8349832B2/en
Application granted granted Critical
Priority to US13/863,103 priority patent/US20130237539A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CANTHERA THERAPEUTICS, INC., ONCOBIOSCIENCES CO. LTD.
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4412Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/80Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D211/84Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen directly attached to ring carbon atoms
    • C07D211/86Oxygen atoms

Definitions

  • the present invention encompasses compounds having the following formula IA, or salts, derivatives, or mixtures thereof:
  • FIG. 1 shows the effect of compounds on pancreatic cells
  • compositions comprising the compounds have utility in the treatment of cancer.

Abstract

The invention relates to compounds and composition for the treatment and prevention of cancer. The invention also covers all diseases that may be treated by selective modulation of levels of reactive oxygen species in diseased cells versus normal cells. Methods for the preparation and administration of such compositions are also disclosed.

Description

CLAIM OF PRIORITY
This application claims priority to U.S. Provisional Application Ser. No. 61/275,754, filed Sep. 2, 2009 and entitled “COMPOUNDS AND COMPOSITIONS FOR TREATING CANCER” which is incorporated by reference in its entirety.”
FIELD OF THE INVENTION
The invention relates to compounds and composition for the treatment and prevention of cancer. The invention also covers all diseases that may be treated by selective modulation of levels of reactive oxygen species in diseased cells versus normal cells. Methods for the preparation and administration of such compositions are also disclosed.
BACKGROUND OF THE INVENTION
The present invention relates to compounds and compositions for the treatment and prevention of cancer, or decreasing the intensity or duration of cancer. The invention also covers all diseases that may be treated by selective modulation of levels of reactive oxygen species in diseased cells versus normal cells.
All living aerobic cells are normally exposed to some reactive oxygen species (ROS) but if ROS levels rise, oxidative stress (OS) occurs, which results in oxygen and oxygen-derived oxidants, and in turn increases the rates of cellular damage. Cells are exposed to both endogenous and exogenous sources of ROS. At high levels, ROS can lead to impaired physiological function through cellular damage of DNA, proteins, lipids, and other macromolecules, which can lead to certain human pathologies including cancers, neurodegenerative disorders, and cardiovascular disease, as well as aging. Moreover, ROS are important in mediating apoptosis.
Selectively increasing levels of ROS in cancer cells and not normal cell may be a safe and effective method for treating cancer. FIG. 6 shows cancer therapies targeting various hallmarks of cancer.
Reducing ROS leads to treatment of neurodegenerative diseases, inflammation and for the treatment of a broad range of disorders including, neurodegenerative diseases, chronic inflammatory diseases, inflammatory bowel disease, rheumatoid arthritis, psoriasis, multiple sclerosis, endotoxin shock, osteoporosis, Alzheimer's disease, congestive heart failure and skin disease.
There remains a need for compounds which can increase ROS in cancer cells while reducing or maintaining ROS levels in normal cells.
SUMMARY OF THE INVENTION
Compounds and compositions that are useful for the treatment and prevention of cancer are provided. The present invention encompasses compounds having the following formula IA, or salts, derivatives, or mixtures thereof:
Figure US08349832-20130108-C00001

or a pharmaceutically acceptable salt or pharmaceutically acceptable derivative thereof, wherein:
  • Ring A is selected from the group consisting of one or more monocyclic aryl, one or more heteroaryl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R1, R2, and R3 is independently selected from the group consisting of hydrogen, halogen, deuterium, CF3, CN, OR, SR, NRR, NRCOR, NRCONRR, NRCO2R, COR, CO2R, NOR, NO2, CONRR, OC(O)NRR, SO2R, SO2NRR, NRSO2R, NRSO2NRR, C(O)C(O)R, or C(O)CH2C(O)R, alkyl, aryl, heteroaryl and morpholino, wherein either R1 and R2, or R2 and R3 are optionally taken together to form a 4-8 membered saturated, partially unsaturated, or fully unsaturated ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur; and z is 0, 1 or 2;
  • each R is independently selected from hydrogen or an optionally substituted C1-C4 aliphatic moiety, wherein:
  • or alternately, two R moieties bound to the same nitrogen atom are optionally taken together with the nitrogen atom to form a 3-7 membered saturated, partially unsaturated, or fully unsaturated ring having 1-2 additional heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • B is selected from:
Figure US08349832-20130108-C00002

wherein
  • R4, R5, R6 and R7 are independently selected from a substituted or unsubstituted C1 to C12 alkyl, a substituted or unsubstituted C1 to C12 alkenyl or a substituted or unsubstituted C1 to C12 alkynyl;
  • X is O, S; and
  • C is a saturated or unsaturated heteroaryl or a saturated or unsaturated C1 to C7 heterocyclic containing one or more hetero atoms wherein the heteroatoms are independently selected from N, O or S;
  • or C a fused ring; and
  • wherein any one or more H is optionally replaced by a deuterium.
The compounds of the present invention may be produced using piperlongumine as a starting material yet offer numerous advantages over piperlongumine. In the course of the experimental investigations, it was found that more than 0.25 mg/ml of piperlongumine in DMSO was precipitated out when diluted 1:10 in phosphate buffered saline or water. It was also found that piperlongumine is soluble in water at 0.1 mg/mL. The compounds of the instant invention are highly soluble in water at concentration of at least 25 mg/mL facilitating pharmaceutical preparation and delivery. Many chemotherapeutic agents transiently increase ROS levels in both cancer cells and normal cells. In contrast, the compounds of the instant invention have a large therapeutic window and do not increase ROS or cause DNA damage in normal cells. Compared to current standards such as etoposide, 5-fluorouracil, cisplatin and taxol, the drug is comparable in its effect on net cancer cell growth (melanoma, ovarian, renal, glioblastoma, drug-resistant non-small cell lung cancer cell lines) as a percent of control.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows the effect of compounds on pancreatic cells
FIG. 2 shows viability of normal breast epithelium in the presence of compounds of the invention.
FIG. 3 shows the effect of compounds on EJ Bladder Carcinoma cells
FIG. 4 shows the effect of compounds on pancreatic cells
FIG. 5 shows the effect of SP2007 and SP83 on bladder tumor xenograft-bearing mice
FIG. 6 shows cancer therapies targeting various hallmarks of cancer.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
Definitions
The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviations, per practice in the art. Alternatively, “about” with respect to the formulations can mean a range of up to 10%, preferably up to 5%.
The terms “alkyl”, “alkenyl”, “alkoxy”, “alkylene”, “alkenylene”, “alkenyl”, “alkyl(arylene)”, “alkynyl”, and “aryl(alkylene)” include, but are not limited to, linear and branched alkyl, alkenyl, alkoxy, alkylene, alkenylene, alkyl(arylene), and aryl(alkylene) groups, respectively.
The phrase “pharmaceutically acceptable” refers to compounds or compositions that are physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a mammal.
An “effective amount of compound” means the amount of compound, salt or salts, or (including its solvates, active metabolites, prodrugs, or racemates or enantiomers thereof (assuming the salt has a chiral center)) that, when administered to a mammal for treating or preventing a state, disorder or condition is sufficient to effect such treatment or prevention. The “effective amount” will vary depending on the active ingredient, the state, disorder, or condition to be treated and its severity, and the age, weight, physical condition and responsiveness of the mammal to be treated. According to one embodiment of the present invention, a therapeutically effective amount of a compound is an amount effective to treat any one of the above mentioned disorders. The compound or its salt or salts may be augmented with a second medication (such as a chemotherapeutic agent, or adjunctive chemotherapeutic agent to treat any of the aforementioned disorders, such as malignancies).
An “effective amount of the pharmaceutical formulation” is an amount of the pharmaceutical formulation described which is effective to treat or prevent a condition in a subject to whom it is administered over some period of time, e.g., provides a therapeutic effect during a desired dosing interval. Generally, an effective amount of the pharmaceutical formulation includes amounts of compound its salt or salts to treat or prevent the desired condition over a desired period of time.
As used herein, the term “treat” includes one or more of the following:
    • (a) arresting, delaying the onset (i.e., the period prior to clinical manifestation of a disorder) and/or reducing the risk of developing or worsening a disorder;
    • (b) relieving or alleviating at least one symptom of a disorder in a mammal, including for example, cancer; or
    • (c) relieving or alleviating the intensity and/or duration of a manifestation of a disorder experienced by a mammal including, but not limited to, those which are in response to a given stimulus (e.g., pressure, tissue injury or cold temperature).
The term “treat” also includes prophylactically preventing, curing, healing, alleviating, relieving, altering, remedying, ameliorating, improving, or affecting a condition (e.g., a disease), the symptoms of the condition, or the predisposition toward the condition.
The term “sustained release” as used herein refers to the release of an active ingredient over an extended period of time leading to lower peak plasma concentrations and a prolonged Tmax as compared to “immediate release” formulations of the same active ingredient.
The term “bioavailability” refers to the rate and extent to which the active ingredient (compound its salt or salts) or active moiety is absorbed from a drug product and becomes systematically available.
The term “polymorph” refers to crystallographically distinct forms of a substance.
The term “hydrate” as used herein includes, but is not limited to, (i) a substance containing water combined in the molecular form and (ii) a crystalline substance containing one or more molecules of water of crystallization or a crystalline material containing free water.
The term “solvate” as used herein includes, but is not limited to, a molecular or ionic complex of molecules or ions of a solvent with molecules or ions of a compound or its salt or salts.
The term “adjunctive chemotherapeutic agent” includes agents which treat, alleviate, relieve, or amelliorate the side effects of chemotherapeutic agents. Such agents include those which modify blood cell growth and maturation. Examples of adjunctive chemotherapeutic agents include, but are not limited to, filgrastim and erythropoietin.
The term “salt” includes compound salt or salts, complexes and active metabolites, prodrugs, racemates, enantiomers, and hydrates thereof.
The term “chemotherapeutic agent” includes any agent which treats, prevents, cures, heals, alleviates, relieves, alters, remedies, ameliorates, improves, or affects malignancies and their metastasis. Examples of such agents (also known as “antineoplastic agents”) include, but are not limited to, prednisone, fluorouracil (e.g., 5-fluorouracil (5-FU)), anastrozole, bicalutamide, carboplatin, cisplatin, chlorambucil, docetaxel, doxorubicin, flutamide, interferon-alpha, letrozole, leuprolide, megestrol, mitomycin, paclitaxel, plicamycin (Mithracin™), tamoxifen, thiotepa, topotecan, valrubicin, vinylastin, vincristine, and any combination of any of the foregoing. Further examples are provided herein.
The term ‘Hydrogen” or “H” includes any form of hydrogen, including deuterium.
Compounds
The present invention provides compounds and compositions which treat and prevent cancer. Compounds of the present invention include those having the following formula I:
Figure US08349832-20130108-C00003

or a pharmaceutically acceptable salt or pharmaceutically acceptable derivative thereof, wherein:
  • Ring A is selected from the group consisting of one or more monocyclic aryl, one or more heteroaryl, a 3-7 membered saturated or partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated, partially unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7 membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered bicyclic saturated or partially unsaturated heterocyclic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen or sulfur, or an 8-10 membered bicyclic heteroaryl ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R1, R2, and R3 independently selected from the group consisting of hydrogen, halogen, deuterium, CF3, CN, OR, SR, NRR, NRCOR, NRCONRR, NRCO2R, COR, CO2R, NOR, NO2, CONRR, OC(O)NRR, SO2R, SO2NRR, NRSO2R, NRSO2NRR, C(O)C(O)R, or C(O)CH2C(O)R, alkyl, aryl, heteroaryl and morpholino, wherein either R1 and R2, or R2 and R3 are optionally taken together to form a 4-8 membered saturated, partially unsaturated, or fully unsaturated ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R is independently selected from hydrogen or an optionally substituted C1-C4 aliphatic moiety (i.e. alkyl, alkenyl, or alkynyl), wherein:
  • or alternately, two R moieties bound to the same nitrogen atom are optionally taken together with the nitrogen atom to form a 3-7 membered saturated, partially unsaturated, or fully unsaturated ring having 1-2 additional heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • B is selected from:
Figure US08349832-20130108-C00004

wherein
  • R4, R5, R6 and R7 are independently selected from a substituted or unsubstituted C1 to C12 alkyl, a substituted or unsubstituted C1 to C12 alkenyl or a substituted or unsubstituted C1 to C12 alkynyl;
  • X is O, S; and
  • C is a saturated or unsaturated heteroaryl or a saturated or unsaturated C1 to C7 heterocyclic containing one or more hetero atoms wherein the heteroatoms are independently selected from N, O or S;
  • or C is a fused ring; and
  • wherein any one or more H is optionally replaced by a deuterium.
In another embodiment, the compounds of the invention are compounds of Formula I above wherein ring A is selected from:
Figure US08349832-20130108-C00005

wherein the ring carries R1, R2 and R3 as defined above;
  • wherein Y is N, O or S; and
  • C is selected from:
Figure US08349832-20130108-C00006

wherein the ring is optionally substituted with one or more R10 and R11, wherein R10 and R11 are independently selected from a substituted or unsubstituted C1 to C12 alkyl, a substituted or unsubstituted C1 to C12 alkenyl or a substituted or unsubstituted C1 to C12 alkynyl, an ether, a thioether, aryl,
    • n is 1, 2 or 3;
    • X1 is O or S;
In a preferred embodiment, the compound of formula I is represented by the following compounds:
Figure US08349832-20130108-C00007
In one embodiment, the compounds of the present invention interact with proteins to increase ROS in cancer cells but not in normal cells. In one embodiment, the compounds of the present invention increase phosphor-JNK levels. In one embodiment, the compounds of the present invention increase p53 activity. In one embodiment, the compounds of the present invention increase p21 activity. In one embodiment, the compounds of the present invention decrease pro-survival gene activity. In one embodiment, the compounds of the present invention induce apoptosis in cancer cell lines.
In another embodiment, the one or more compounds of the instant invention are used to treat cancer. Another embodiment includes a method for increasing apoptosis by administering a compound of the instant invention. Another embodiment includes a method for increasing p53 activity by administering one or more compounds of the instant invention. Another embodiment includes a method for increasing p21 activity by administering one or more compounds of the instant invention. Another embodiment includes a method of preferentially inducing DNA damage in cancer cells by administering one or more compounds of the instant invention. Another embodiment is a method of suppressing DNA damage in normal cells by administering one or more compounds of the instant invention.
In another embodiment, the invention provides a method for improving the expression of proteins associated with normal cell survival. In another embodiment, the invention provides for a method of sparing normal cells while killing cancer cells.
In another embodiment, the invention provides a method of administering a compound of the instant invention at a dose of about 100 mg/kg to about 3000 mg/kg. In another embodiment the invention provides a method of administering a compound of the instant invention at a dose of about 2.5 mg/kg to about 10 mg/kg. In some embodiments, a therapeutically effective amount is less than 50 mg/kg, such as less than 45 mg/kg, less than 40 mg/kg, less than 35 mg/kg, less than 30 mg/kg, less than 25 mg/kg, less than 20 mg/kg or less than 15 mg/kg. In some embodiments, a therapeutically effective amount is less than 10 mg/kg, such as less than 9 mg/kg, less than 8 mg/kg, less than 7 mg/kg, less than 6 mg/kg, less than 5 mg/kg, less than 4 mg/kg, less than 3 mg/kg or less than 2 mg/kg. In some embodiments, a therapeutically effective amount is less than 1.5 mg/kg, such as less than 1.4 mg/kg, less than 1.3 mg/kg, less than 1.2 mg/kg, less than 1.1 mg/kg, less than 1 mg/kg, less than 0.9 mg/kg, less than 0.8 mg/kg, less than 0.7 mg/kg, less than 0.6 mg/kg, less than 0.5 mg/kg, less than 0.4 mg/kg, less than 0.3 mg/kg, less than 0.2 mg/kg or less than 0.1 mg/kg.
In another aspect, the invention provides a method for inhibiting cell proliferation. In some embodiments, the method for inhibiting cell proliferation comprises contacting a cell with an effective amount of a composition comprising a compound of the invention to inhibit the proliferation of the cell. In some embodiments, the method further comprises contacting the cells with a compound of the invention and a chemotherapeutic agent. In another aspect, the invention provides a method for increasing apoptosis of a cell or in a population of cells. In some embodiments, the method for increasing apoptosis of a cell or in a population of cells, the method comprises contacting the cell or population of cells with an effective amount of a composition comprising a compound of the invention to increase apoptosis in the cell or population of cells. In some embodiments, the number of apoptotic cells in a population of cells is increased by at least two-fold. In some embodiments, the number of apoptotic cells in a population of cells is increased by at least five-fold. In some embodiments, the number of apoptotic cells in a population of cells is increased by at least ten-fold. In some embodiments, the method further comprises contacting the cells with a compound of the invention and a chemotherapeutic agent.
In another embodiment one or more compounds of the instant invention are administered with one or more chemotherapeutic agent. In another embodiment, one or more compounds of the instant invention are administered prior to one or more chemotherapeutic agents. In another embodiment, one or more compounds of the instant invention are administered following one or more chemotherapeutic agents.
Chemotherapeutic agents include, but are not limited to an agent which is administered to a subject for the purpose of treating a cancer. Chemotherapeutic agents include, but are not limited to antiproliferative compounds, anti-neoplastic compounds, anti-cancer supplementary potentiating agents and radioactive agents. One of ordinary skill in the art is familiar with a variety of chemotherapeutic agents, or can find those agents in the routine art, which are used in the medical arts to treat cancer.
Chemotherapeutic agents include, but are not limited to, the following sub-classes of compounds: Antineoplastic agents such as: Acivicin; Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Adriamycin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Buniodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer; Carboplatin; Carmustine; Carubicin Hydrochloride; Carzelesin; Cedefingol; Chlorombucil; Cirolemycin; Cisplatin; Cladribine; Crisnatol Mesylate; Cyclophosphamide; Cytarabine; Dacarbazine; DACA (N-[2-(Dimethyl-amino)ethyl]acridine-4-carboxamide); Dactinomycin; Daunorubicin Hydrochloride; Daunomycin; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine Ifesylate; Diaziquone; Docetaxel; Doxorubicin; Doxorubicin Hydrochloride; Droloxifene; Droloxifene Citrate; Dromostanolone Propionate; Duazomycin; Edatrexate; Eflornithine Hydrochloride; Elsamitrucin; Enloplatin; Enpromate; Epipropidine; Epirubicin 30 Hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Estramustine Phosphate Sodium; Etanidazole; Ethiodized Oil I 131; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole Hydrochloride; Fazarabine; Fenretinide; Floxuridine; Fludarabine Phosphate; Fluorouracil; 5-FdUMP; Fluorocitabine; Fosquidone; Fostriecin Sodium; Gemcitabine; Gemcitabine Hydrochloride; Gold Au 198; Hydroxyurea; Idarubicin Hydrochloride; Ifosfamide; Ilmofosine; Interferon Alfa-2a; Interferon Alfa-2b; Interferon Alfa-n1; Interferon Alfa-n3; Interferon Beta-1a; Interferon Gamma-1b; Iproplatin; Irinotecan Hydrochloride; Lanreotide Acetate; Letrozole; Leuprolide Acetate; Liarozole Hydrochloride; 5 Lometrexol Sodium; Lomustine; Losoxantrone Hydrochloride; Masoprocol; Maytansine; Mechlorethamine Hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Methotrexate Sodium; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitocromin; Mitogillin; Mitomalcin; Mitomycin, Mitosper; Mitotane; Mitoxantrone Hydrochloride; Mycophenolic Acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel Pegaspargase; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Plicamycin; Plomestane; Porfimer Sodium; Porfiromycin; Prednimustine; Procarbazine Hydrochloride; Puromycin; Puromycin Hydrochloride; Pyrazofurin; Riboprine; Rogletimide; Safingol; Safingol Hydrochloride; Semustine; Simtrazene; Sparfosate Sodium; Sparsomycin; Spirogermanium Hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Streptozocin; Strontium Chloride Sr 89; Sulofenur; Talisomycin; Taxane; Taxoid; Tecogalan Sodium; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Thymitaq; Tiazofurin; Tirapazamine; Tomudex; TOP-53; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Triptorelin; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Verteporfin; Vinblastine; Vinblastine Sulfate; Vincristine; Vincristine Sulfate, Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglycinate Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate; Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; Zorubicin Hydrochloride; 2-Chlorodeoxyadenosine; 2′-Deoxyformycin; 9-aminocamptothecin; raltitrexed; N-propargyl-5,8-dideazafolic acid, 2-chloro-2′-arabino-fluoro-2′-deoxyadenosine; 2-chloro-2′-deoxyadenosine; anisomycin; trichostatin A; hPRL-G129R; CEP-751; linomide; Piritrexim Isethionate; Sitogluside; Tamsulosin Hydrochloride and Pentomone. Anti-neoplastic compounds include, but are not limited to 20-epi-1,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antogonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives (e.g., 10-hydroxy-camptothecin); canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorins; chloroquinoxaline sulfonamide; cicaprost; 15 cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin 13; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin 10 deslorelin; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; dihydrotaxol; dioxamycin; diphenyl spiromustine; discodermolide; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epothilones; epithilones; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide; etoposide 4′-phosphate (etopofos); exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol; irinotecan; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mithracin; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistancegene inhibitor, multiple tumor suppressor 1-based therapy; mustard anticancer agent; 15 mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinum-triamine complex; podophyllotoxin; porfimer sodium; porfiromycin; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; Sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen binding protein; sizofuran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; 5 sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thalidomide; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene dichloride; topotecan; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; zinostatin stimalamer. Anti-cancer supplementary potentiating agents include, but are not limited to, Tricyclic anti-depressant drugs (e.g., imipramine, desipramine, amitryptyline, clomipramine, trimipramine, doxepin, nortriptyline, protriptyline, amoxapine and maprotiline); non-tricyclic anti-depressant drugs (e.g., sertraline, trazodone and citalopram); Ca2+ antagonists (e.g., verapamil, nifedipine, nitrendipine and caroverine); Calmodulin inhibitor (e.g. prenylamine, trifluoroperazine and clomipramine); Amphotericin B; Triparanol analogues (e.g. tamoxifen); antiarrhythmic drugs (e.g., quinidine); antihypertensive drugs (e.g. reserpine); Thiol depleters (e.g., buthionine and sulfoximine) and Multiple Drug Resistance reducing agents such as Cremaphor EL. The compounds of the invention also can be administered with cytokines such as granulocyte colony stimulating factor.
Radioactive agents include but are not limited to Fibrinogen I 125; Fludeoxyglucose F18; Fluorodopa F 18; Insulin I 125; Insulin I 131; Iobenguane I 123; Iodipamide Sodium I 131; Iodoantipyrine I 131; Iodocholesterol I 131; Iodohippurate Sodium I 123; Iodohippurate-23-Sodium I 125; Iodohippurate Sodium I 131; Iodopyracet I 125; Iodopyracet I 131; Iofetamine Hydrochloride I 123; Iomethin I 125; Iomethin I 131; Iothalamate Sodium I 125; Iothalamate Sodium I 131; Iotyrosine I 131; Liothyronine I 125; Liothyronine I 131; Merisoprol Acetate Hg 197; Merisoprol Acetate-Hg 203; Merisoprol Hg 197; Selenomethionine Se 75; Technetium Tc 99m Atimony Trisulfide Colloid; Technetium Tc 99m Bicisate; Technetium Tc 99m Disofenin; Technetium Tc 99m Etidronate; Technetium Tc 99m Exametazime; Technetium Tc 99m Furifosmin; Technetium Tc 99m Gluceptate; Technetium 99m Lidofenin; Technetium Tc 99 mm Mebrofenin; Technetium Tc 99m Medronate; TechnetiumTc 99m Medronate Disodium; Technetium Tc 99m Mertiatide; Technetium Tc 99m Oxidronate; Technetium Tc 99m Pentetate; Technetium Ic 99m Pentetate Calcium Trisodium; Technetium Tc 99m Sestamibi; Technetium Tc 99m Siboroxime; Technetium Tc 99m Succimer; Technetium Tc 99m Sulfur Colloid; Technetium Tc 99m Teboroxime; Technetium Tc 99m Tetrofosmin; Technetium Tc 99m Tiatide; Thyroxine I 125: Thyroxine I 131; Tolpovidone I 131; Triolein I 125; Triolein I 131.
The compounds of the instant invention are efficacious in the treatment and/or prevention of a broad range of cancers. Such cancers include but are not limited to lung cancer, bladder cancer, melanoma, bladder carcinoma, pancreatic cancer, breast cancer.
In one aspect, the invention provides methods for the treatment of cancer. “Cancer” as used herein refers to an uncontrolled growth of cells which interferes with the normal functioning of the bodily organs and systems. Cancers which migrate from their original location and seed vital organs can eventually lead to the death of the subject through the functional deterioration of the affected organs. Carcinomas are malignant cancers that arise from epithelial cells and include adenocarcinoma and squamous cell carcinoma. Sarcomas are cancer of the connective or supportive tissue and include osteosarcoma, chondrosarcoma and gastrointestinal stromal tumor. Hematopoietic cancers, such as leukemia, are able to outcompete the normal hematopoietic compartments in a subject, thereby leading to hematopoietic failure (in the form of anemia, thrombocytopenia and neutropenia) ultimately causing death. A person of ordinary skill in the art can classify a cancer as a sarcoma, carcinoma or hematopoietic cancer. Cancer, as used herein, includes the following types of cancer, breast cancer, biliary tract cancer; bladder cancer; brain cancer including glioblastomas and medulloblastomas; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms including acute lymphocytic and myelogenous leukemia; T-cell acute lymphoblastic leukemia/lymphoma; hairy cell leukemia; chromic myelogenous leukemia, multiple myeloma; AIDS-associated leukemias and adult T-cell leukemia lymphoma; intraepithelial neoplasms including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer including squamous cell carcinoma; ovarian cancer including those arising from epithelial cells, stromal cells, germ cells and mesenchymal cells; pancreatic cancer; prostate cancer; rectal cancer; sarcomas including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma, and osteosarcoma; skin cancer including melanoma, Kaposi's sarcoma, basocellular cancer, and squamous cell cancer; testicular cancer including germinal tumors such as seminoma, non-seminoma (teratomas, choriocarcinomas), stromal tumors, and germ cell tumors; thyroid cancer including thyroid adenocarcinoma and medullar carcinoma; and renal cancer including adenocarcinoma and Wilms tumor. Other cancers will be known to one of ordinary skill in the art.
The compounds may be in the form of the carboxylic acid and/or their salts. Salts include but are not limited to organic and inorganic salts, for example alkali-metal salts, such as sodium, potassium and lithium; alkaline-earth metal salts, such as magnesium, calcium or barium; ammonium salts; basic amino acids such as lysine or arginine; and organic amines, such as dimethylamine or pyridine. Preferably, the salts are tartrate and will be hydrochloride salts. The salts may be mono- or multi-valent salts, such as monosodium salts and di-sodium salts. The salts may also be solvates including ethanol solvates.
The compounds described herein can be readily prepared synthetically by methods described herein. For example, the compounds may be prepared as in Example 1. Other methods of preparation are known to those skilled in the art and are found, for example in Synthesis of diverse analogues of Oenostacin and their antibacterial activities (Vandana, Bioorganic & Medicinal Chemistry 15 (2007) 518-525); A Simple Regioselective Demethylation of p-Aryl Methyl Ethers Using Aluminum Chloride-Dichloromethane System (Negi, Synthetic Communications, 35: 15-21, 2005); Antiplatelet Activities of Newly Synthesized Derivatives of Piperlongumine (Park, Phytother. Res. 22, 1195-1199 (2008)); On the structure of Pipertine and a synthesis of dihydropiplartine (Joshi, Tetrahedron Letters No. 20, pp. 2395-2420, 1968); Synthesis and Molecular Structure of Piplartine (Boll, Tetrahedron Letters No. 40, pp. 171-175, 1984); Isolation, Synthesis and Evolutionary Ecology of Piper Amides: June 2004 (Dyer); Inter- and Intraspecific Compararisons of Antiherbivore Defenses in Three Species of Rainforest Understory Shrubs (Fincher, J Chem Ecol (2008) 34: 558-557). The contents of these references and these methods are incorporated by reference in their entirety.
Salts of the present compound may be made by methods known in the art. For example, sodium salts may be made by dissolving the compound in ethanol and adding aqueous sodium hydroxide. Hydrochloride and Tartrate salts are prepared as described in Examples 1 and 2.
Scheme I represents the preparation of Compound 1 and its tartaric acid salt.
Figure US08349832-20130108-C00008
Another embodiment is a method of preparation of the compounds of the instant invention. In yet another embodiment, the preparation of the representative compounds of the instant invention is represented by Scheme II:
Figure US08349832-20130108-C00009
Mechanism
Without being bound to any particular theory, the compounds of the present invention exert their therapeutic effect by reducing oxidative stress and thereby reducing damage to DNA in normal cells while increasing ROS levels in cancer cells. The resulting effect of exposure to Reactive oxygen species (ROS) is often changes in cell differentiation and finally, over a prolonged period, apoptosis. Under non-stressed conditions, GSTP1 inhibits JNK phosphorylation by sequestering the JNK-c-JUN complex. Stressor triggers oligomeraization of GSTP1 and results in dissociation of the JNK-c-JUN complex. JNK can then become phosphorylated and activate downstream kinase and transcription factors.
Reactive oxygen species (ROS) have been shown to be increased in, for example, EJ Bladder Carcinoma cells, Breast Carcinoma cells, and removal of GSTJI has been shown to block the increase in ROS. SP2007 has been shown to increase ROS in cancer cells and causes apoptosis. Other compounds of this invention have been shown to be efficacious in vivo against pancreatic cancer cells, breast epithelium and EJ Bladder cancer cells.
Furthermore, compounds of the instant invention are superior to the traditional Piper longum parent molecule in that the compounds are highly soluble. For example whereas the parent Piper longum can dissolve only in quantities of 0.1 mg/ml in water, much larger quantities of compounds of the present invention are soluble in water. For example, the tartrate salt of compound one of the present invention is soluble at 100 mg/ml of water.
Pharmaceutical Forms
The administration compositions may alternately be in the form of a solid, such as a tablet, capsule or particle, such as a powder or sachet. Solid dosage forms may be prepared by mixing the solid form of the compound with the solid form of the active agent. Alternately, a solid may be obtained from a solution of compound and active agent by methods known in the art, such as freeze drying, precipitation, crystallization and solid dispersion.
The amount of compound used in an administration composition of the present invention is an amount effective to accomplish the purpose of the particular compound. However, the amount can be less than that amount when the composition is used in a dosage unit form because the dosage unit form may contain a plurality of compound compositions or may contain a divided pharmacologically, biologically, therapeutically, or chemically effective amount. The total effective amount can then be administered in cumulative units containing, in total, an effective amount of the active agent.
The total amount of compound to be used can be determined by methods known to those skilled in the art.
The presently disclosed compounds may be in any dosage form known to those skilled in the art, particularly suitable for oral, intranasal, sublingual, intraduodenal, subcutaneous, buccal, intracolonic, rectal, vaginal, mucosal, pulmonary, transdermal, intradermal, parenteral, intravenous, intramuscular, intrathecal, intraperitoneal and ocular administrations, as well as traversing the blood-brain barrier. The compounds may also be prepared for injections, either by infusion into a vein or artery or injected into the peritoneum, bladder, or directly into tumors. Topical preparations such as creams, ointments, gels, lotions or transdermal patches are contemplated dosage forms.
Dosage unit forms can also include any one or combination of excipients, diluents, disintegrants, lubricants, plasticizers, colorants, flavorants, taste-masking agents, sugars, sweeteners, salts, and dosing vehicles, including, but not limited to, water, 1,2-propane diol, ethanol, olive oil, or any combination thereof.
The compounds and compositions of the subject invention are useful for administering biologically or chemically active agents to any animals, including but not limited to birds such as chickens; mammals, such as rodents, cows, pigs, dogs, cats, primates, and particularly humans; and insects.
The compounds of the invention can be administered to animal and man, directly or together with a vehicle commonly used. The dose form is not particularly limited, and is selected appropriately as needed on use, including oral drugs such as tablets, capsules, granules, grains and powder, or non-oral drugs such as injection and suppository.
The oral drugs are prepared by ordinary methods, using starch, lactose, sucrose, mannitol, carboxymethylcellulose, corn starch, or inorganic salts.
In the drugs of this type, use can be made of a binder, a disintegrator, a surfactant, a lubricant, a fluidity-promoting agent, a flavor, a tinction, a perfume and the like, in addition to the vehicle mentioned above. Non-limiting examples of some of these substances are presented below:
Binder: Starch, dextrin, powdered acasia, gelatin, hydroxypropylstarch, methylcellulose, carboxymethylcellulose sodium, hydroxypropylcellulose, crystalline cellulose, ethylcellulose, polyvinylpyrrolidone, and macrogol.
Disintegrator: Starch, hydroxypropylstarch, carboxymethylcellulose sodium, carboxymethylcellulose calcium, carboxymethylcellulose, and low-substituted hydroxypropylcellulose.
Surfactant: Sodium laurylsulfate, soybean lecithin, succharose fatty acid ester, and polysorbate 80.
Lubricant: Talc, waxes, hydrogenated vegetable oil, succharose fatty acid ester, magnesium stearate, calcium stearate, aluminum stearate, and polyethylene glycol.
Fluidity-Promoting Agent: Light anhydrous silicic acid, dry aluminum hydroxide gel, synthetic aluminum silicate, and magnesium silicate.
The compounds of the present invention can also be administered in the form of suspension, emulsion, syrup or elixir. These forms of drugs may also contain flavor, perfume and tinction.
The non-oral drugs can be prepared by ordinary methods, and use may be made of an attenuant such as distilled water for injection, physiological saline, aqueous solution of glucose, vegetable oil for injection, sesame oil, peanut oil, soybean oil, corn oil, propylene glycol, or polyethylene glycol. If necessary, germicide, preservative, and stabilizer may be added. The non-oral drugs can be filled in a vial or the like and frozen, and then be removed of water by the ordinary freeze-dry technique. A liquid drug can be reformulated from the freeze-dried drug immediately before administration. Further, an isotonic, a stabilizer, a preservative, an antiseptic, a sedative, and the like may be added.
The compositions comprising the compounds have utility in the treatment of cancer.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The following examples illustrate the invention without limitation. All parts are given by weight unless otherwise indicated.
EXAMPLE 1
Compound Preparation
1a. Preparation of Compound 1 and its Tartaric Acid Salt.
Scheme I represents the preparation of Compound 1 and its tartaric acid salt.
Figure US08349832-20130108-C00010

Preparation of BPS-02083-01
To a stirred solution of Piperlongumine (5.60 g, 17.6 mmol) in anhydrous DCM was added AlCl3 (23.3 g, 174 mmol) at 28° C. AlCl3 cannot be completely dissolved in the solution and the red suspension became a yellow suspension while stirred at 28° C. for 15 min. Saturated NaHCO31 was added and the mixture was extracted with dichloromethane (150 mL*2). The combined organic layers were washed with brine (50 mL*3), dried over anhydrous Na2SO4, concentrated in vacuo and washed with dichloromethane/petroleum ether (v/v=2:3) to give BPS-02083-01 (3.5 g, 11.6 mmol, 66%) as a yellow solid. MS (ESI/FT-MS): 304 [M+H]+; expected 304.
Preparation of Compound I
To a stirred solution of BPS-02083-01 intermediate (1.94 g, 6.4 mmol), 2-morpholinoethanol (1.0 g, 7.7 mmol) and PPh3 (2.52 g, 9.6 mmol) in anhydrous dichloromethane (80 mL) was added slowly a solution of DEAD (1.67 g, 9.6 mmol) in dichloromethane (20 mL) under N2 at 0° C. The mixture was slowly warmed to room temperature and stirred overnight. The solvent was removed in vacuo and purified by chromatography on silica gel (petroleum ether/ethyl acetate=1:1 to 2:1) to give BPS-02083-00 (1.4 g, 3.37 mmol, 53%) as a pale yellow oil. MS (ESI/FT-MS): 417 [M+H]+; expected 417.
Compound 1-L-tartaric acid salt.
To a solution of BPS-02083-00 [Compound 1] (4.16 g, 10.0 mmol) in dichloromethane (25 mL) was added a solution of L-tartaric acid (1.50 g, 10.0 mmol) in dichloromethane/methanol (10 mL). The solvent was removed in vacuo to give the L-tartaric acid salt of BPS-02083-00 (5.66 g, 10.0 mmol, 100%) as a colorless solid. MS (ESI/FT-MS): 417 [M+H]+; expected 417, same as the free base.
EXAMPLE 2
Attempted Preparation of Compound I HCL Salt
5 gram free base of Compound I (thick oil) (˜97% by HPLC). BPS-02083-00 (100 mg) was dissolved in ether (2.0 mL) and cooled to −10° C. HCl in ether (saturated) was added and stirred for 10 min. The solvent was removed (<25° C.) to give a colorless solid (BPS-02083-00-HCl). The spectra indicated that it is not the HCl salt of BPS-02083-00. The structure is B (HCl addition to double bond).
Figure US08349832-20130108-C00011
EXAMPLE 3
Preparation of Representative Compounds of the Invention
The Intermediate was prepared as in Example 1 and the procedure for the de-boc step to form the by product is as follows. To a solution of BPS-02085 (about 35 mg) in menthol (5.0 ml) was bubled HCl(g) for 15 minutes at room temperature (20° C.). The solvent was removed in vacuo and the residue was purified by TLC (DCM:MA:NH3H2O=70:4:1) to give by-product (confirmed by 1NMR and LCMS). Non-limiting example of compounds which may be prepared by the ad-boc method described in Example 3 are represented below.
Figure US08349832-20130108-C00012
EXAMPLE 4
Report of Representative Compounds
SM Quantity Product Quantity HPLC HNMR LCMS[M + H]+
SM-00 190 mg BPS- 124 mg + 32 mg >95% Passed 304
02084-01 SM-00
SM-00 9.3 mg BPS- 254 mg + 84 mg >95% Passed 304
SM-00 13 mg 02084-01 SM-00 304
SM-00 176 mg 304
SM-00 190 mg 304
BPS- 56 mg BPS- 7.1 mg 95.0%  Passed 417
02084-01 02083-00
BPS- 56 mg BPS- 22 mg 96.1%  Passed 375
02084-01 02084-00
BPS- 96 mg BPS- 30-40 mg >90% Passed
02084-01 02085-01
BPS- 30-40 mg BPS- 0 mg
02085-01 02085-00
BPS- 65 mg + 36 mg BPS- 25-30 mg >90% Passed
02084-01 02086-01
BPS- 25-30 mg BPS- 0 mg
02086-01 02085-00
BPS- 56 mg BPS- 42 mg 97.1%  Passed 332
02084-01 02087-00
EXAMPLE 5
Effect of Compound on Pro-Survival Gene Suppression
Additional studies will demonstrate that compounds of the instant invention have a large therapeutic window by sparing normal cells, such as fibroblasts and keratocytes, but killing tumors cells, such as EJ cells and HCT116 cells. Studies will also demonstrate that DNA in normal cells remains unaffected by compounds of the instant invention and that there is no increase in ROS in normal cells.
FIGS. 1-4 show in-vivo efficacy of the compounds against pancreatic cancers cells, breast epithelium and EJ Bladder Cancer Cells.
EXAMPLE 6
Antitumor Effect of SP2007 and SP830n Bladder Tumor Xenograft-Bearing Mice
We tested the anti-tumor effect SP2007 and SP83 in bladder tumor xenograft-bearing mice.
Four nude/nude mice weighing approximately 0.02 Kg were fasted overnight and anestatized with Avertin (2,2,2, Tribromoethanol), 250 mg/kg, body weight; 1P injection. A total 2×106 EJ cells (acquired from Japanese cellbank) were implanted subcutaneously on opposite site flanks in each of four nude/nude mice in each treatment group. Mice were fed and given water ad lib. When tumor masses grew to approximately 2 to 5 mm in diameter, control vehicle, SP2007 or SP83 were administered intraperitoneally (total 2 mg/kg) every 24 hours for 25 days. Anti-tumor effects were observed in SP2007 and SP83 administered tumor mice, as compared to control DMSO-administered tumor mice. Results are shown in FIG. 5.
Although this invention has been described with a certain degree of particularity, it is to be understood that the present disclosure has been made only by way of illustration and that numerous changes in the details of construction and arrangement of parts may be resorted to without departing from the spirit and the scope of the invention. Many variations of the present invention will suggest themselves to those skilled in the art in light of the above detailed description. All such obvious variations are within the fully intended scope of the appended claims.

Claims (12)

1. A compound or a pharmaceutically acceptable salt thereof, wherein the compound is selected from:
Figure US08349832-20130108-C00013
2. A pharmaceutical composition comprising one or more compounds of claim 1 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
3. The pharmaceutical composition of claim 2, further comprising one or more chemotherapeutic agents.
4. A kit comprising a pharmaceutical composition comprising a compound of claim 1 or a pharmaceutically acceptable salt thereof, and instructions for preparation and/or administration of the pharmaceutical composition.
5. The kit of claim 4, further comprising one or more chemotherapeutic agents.
6. A method for, delaying an onset of, reducing a risk of developing or worsening of, relieving or alleviating a symptom of, or relieving or alleviating an intensity or duration of a manifestation of a cancer in a subject, the method comprising: administering to a subject in need thereof a compound of claim 1 or a pharmaceutically acceptable salt thereof, wherein the cancer is selected from the group consisting of a pancreatic cancer, a breast cancer, and a bladder cancer.
7. The method of claim 6, comprising inhibiting further growth of the cancer, wherein the cancer is selected from the group consisting of a pancreatic cancer, a breast cancer, and a bladder cancer.
8. The method of claim 6 comprising administering to the subject from about 2.5 mg/kg to about 100 mg/kg of a compound of claim 3, or a pharmaceutically acceptable salt thereof.
9. A method for inhibiting cell proliferation, the method comprising: contacting a cell with a composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, to inhibit the proliferation of the cell, wherein the cell is selected from the group consisting of a pancreatic cancer cell, a breast cancer cell, and a bladder cancer cell.
10. A method for increasing apoptosis of a cell or in a population of cells, the method comprising: contacting the cell or population of cells with a composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, to increase apoptosis in the cell or population of cells, wherein the cell or population of cells is selected from the group consisting of a pancreatic cancer cell, a population of pancreatic cancer cells, a breast cancer cell, a population of breast cancer cells, a bladder cancer cell, and a population of bladder cancer cells.
11. A method for increasing p53 activity in a cell or population of cells, the method comprising: contacting the cell or population of cells with a composition comprising a compound of claim 1, or a pharmaceutically acceptable salt thereof, to increase p53 activity in the cell or population of cells.
12. The compound of claim 1, wherein said pharmaceutically acceptable salt is selected from tartrate and hydrochloride.
US12/874,306 2009-09-02 2010-09-02 Compounds and compositions for treating cancer Active US8349832B2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US12/874,306 US8349832B2 (en) 2009-09-02 2010-09-02 Compounds and compositions for treating cancer
PCT/US2011/025022 WO2012030408A1 (en) 2010-09-02 2011-02-16 Compounds and compositions for treating cancer
US13/028,571 US8318737B2 (en) 2009-09-02 2011-02-16 Compounds and compositions for treating cancer
US13/162,632 US20120157455A1 (en) 2009-09-02 2011-06-17 Compounds And Compositions For Treating Cancer
US13/679,522 US20130203757A1 (en) 2009-09-02 2012-11-16 Compounds And Compositions For Treating Cancer
US13/863,103 US20130237539A1 (en) 2009-09-02 2013-04-15 Compounds and Compositions for Treating Cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US27575409P 2009-09-02 2009-09-02
US12/874,306 US8349832B2 (en) 2009-09-02 2010-09-02 Compounds and compositions for treating cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/028,571 Continuation-In-Part US8318737B2 (en) 2009-09-02 2011-02-16 Compounds and compositions for treating cancer

Publications (2)

Publication Number Publication Date
US20110053938A1 US20110053938A1 (en) 2011-03-03
US8349832B2 true US8349832B2 (en) 2013-01-08

Family

ID=45774359

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/874,306 Active US8349832B2 (en) 2009-09-02 2010-09-02 Compounds and compositions for treating cancer

Country Status (2)

Country Link
US (1) US8349832B2 (en)
WO (1) WO2012030408A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140024639A1 (en) * 2012-07-20 2014-01-23 The Broad Institute, Inc. Compounds, Compositions, and Methods for Cancer Therapy
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9801922B2 (en) 2011-08-03 2017-10-31 University Of Iowa Research Foundation Compositions and methods of treating cancer
WO2014032019A2 (en) * 2012-08-23 2014-02-27 Georgetown University Compounds and methods of use thereof for treating tumors
EP3708170A1 (en) 2014-05-05 2020-09-16 BioVentures, LLC Compositions and methods for inhibiting antiapoptotic bcl-2 proteins as anti-aging agents
CA2955972A1 (en) 2014-07-22 2016-01-28 Bioventures, Llc. Compositions and methods for selectively depleting senescent cells
EP3328372A4 (en) 2015-07-28 2019-03-20 University Of Iowa Research Foundation Compositions and methods of treating cancer
CA3018991A1 (en) 2016-04-21 2017-10-26 Bioventures, Llc Compounds that induce degradation of anti-apoptotic bcl-2 family proteins and the uses thereof
EP3474879A4 (en) 2016-06-24 2020-05-06 University of Iowa Research Foundation Compositions and methods of treating melanoma
WO2019075367A1 (en) 2017-10-13 2019-04-18 Tolero Pharmaceuticals, Inc. Pkm2 activators in combination with reactive oxygen species for treatment of cancer
WO2019221755A1 (en) * 2018-05-18 2019-11-21 Bioventures, Llc Piperlongumine analogues and uses thereof
US11529335B2 (en) 2020-07-31 2022-12-20 University Of Iowa Research Foundation Compositions and methods for treating cancer
WO2023114455A1 (en) * 2021-12-17 2023-06-22 University Of Florida Research Foundation, Incorporated Discovery of piperlongumine as a novel e3 ligase ligand

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6683079B2 (en) 1998-04-27 2004-01-27 Centre National De La Recherche Scientifique 3-(amino-or aminoalkyl) pyridinone derivatives and their use for the treatment of HIV related diseases
US6706021B2 (en) 1997-11-26 2004-03-16 3-Dimensional Pharmaceuticals, Inc. Heteroaryl aminoguanidines and alkoxyguanidines and their use as protease inhibitors
US20050250768A1 (en) 1998-12-29 2005-11-10 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory and immune-suppressive compounds
WO2006047354A1 (en) 2004-10-26 2006-05-04 Novartis Ag Pyrrolo [1,2-d][1,2-4]triazine as inhibitors of c-jun n terminal kinases (jnk) and p-38 kinases
US20070244164A1 (en) 2006-04-18 2007-10-18 Ardea Biosciences Pyridone sulfonamides and pyridone sulfamides as mek inhibitors
US20080220416A1 (en) 2004-05-20 2008-09-11 The Board Of Trustees Of The University Of Illinoi Compositions for Inhibiting Cell Growth and Inducing Apoptosis in Cancer Cells and Methods of Use Thereof
WO2008112195A1 (en) 2007-03-09 2008-09-18 Bioseek, Inc. Methods for identifying agents and their use for the prevention or stabilization of fibrosis
US20080227829A1 (en) 2007-02-14 2008-09-18 Hammerstone John F Neurogenic compounds
US20090312373A1 (en) 2008-03-11 2009-12-17 The General Hospital Corporation Methods for the treatment of cancer using piperlongumine and piperlongumine analogs

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6706021B2 (en) 1997-11-26 2004-03-16 3-Dimensional Pharmaceuticals, Inc. Heteroaryl aminoguanidines and alkoxyguanidines and their use as protease inhibitors
US6683079B2 (en) 1998-04-27 2004-01-27 Centre National De La Recherche Scientifique 3-(amino-or aminoalkyl) pyridinone derivatives and their use for the treatment of HIV related diseases
US20050250768A1 (en) 1998-12-29 2005-11-10 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory and immune-suppressive compounds
US20080220416A1 (en) 2004-05-20 2008-09-11 The Board Of Trustees Of The University Of Illinoi Compositions for Inhibiting Cell Growth and Inducing Apoptosis in Cancer Cells and Methods of Use Thereof
WO2006047354A1 (en) 2004-10-26 2006-05-04 Novartis Ag Pyrrolo [1,2-d][1,2-4]triazine as inhibitors of c-jun n terminal kinases (jnk) and p-38 kinases
US20070244164A1 (en) 2006-04-18 2007-10-18 Ardea Biosciences Pyridone sulfonamides and pyridone sulfamides as mek inhibitors
US20080227829A1 (en) 2007-02-14 2008-09-18 Hammerstone John F Neurogenic compounds
WO2008112195A1 (en) 2007-03-09 2008-09-18 Bioseek, Inc. Methods for identifying agents and their use for the prevention or stabilization of fibrosis
US20090312373A1 (en) 2008-03-11 2009-12-17 The General Hospital Corporation Methods for the treatment of cancer using piperlongumine and piperlongumine analogs

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
Bezarra et al.; Research Thesis; Estudo das Propriedades Anticancer da Piplartina; Jun. 27, 2008.
Bezerra, D.P., et al; In vivo growth-inhibition of Sarcoma 180 by piplartine and piperine, two alkaloid amides from Piper; Brazilian Journal of Medical and Biological Research (2006) 39:801-807.
Boll, et al., "Synthesis and Molecular Structure of Piplartine", Tetrahedron vol. 40, No. 1, pp. 171-175, (1984).
Dyer, et al., "Isolation, Synthesis and Evolutionary Ecology of Piper Amides", PI: MRM/IKJ, 117-139, 2004.
Fincher, et al. , "Inter- and Intraspecific Comparisons of Antiherbivore Defenses in Three Species of Rainforest Understory Shrubs", J Chem Ecol, vol. 34, pp. 558-574, (2008).
Joshi, et al., On the Structure of Piplartine and a Synthesis of Dipydropiplartine, Tetrahedron Letters No. 20, Pergamon Press, pp. 2395-2400, (1968).
Negi, et al. "A Simple Regioselective Demethylation of p-Aryl Methyl Ethers Using Aluminum Chloride-Dicholoromethane System", Synthetic Communications, vol. 35:1,pp. 15-21, 2005.
Park et al., caplus an 2008:1229445. *
Park, et al. "Antiplatelet Activities of Newly Synthesized Derivatives of Piperlongumine", Phythother.Res.22, pp. 1195-1999, (2008).
Proghost Research, "Engines of Momentum: Clinical Trial News: TELIK (TELK)" Dec. 6, 2004 1-2.
Raza, et al., "Phase 1-2a multicenter dose-escalation study of ezatiostat hydrochloride liposomes for injection" J. Hematalogy & Oncology, 2:20, May 13, 2009; 12 total pages.
Srivastava et al., "Synthesis of diverse analogues of Oenostacin and their antibacterial activities", Biorganic & Medicinal Chemistry, vol. 15; Oct. 10, 2006, pp. 518-525.

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140024639A1 (en) * 2012-07-20 2014-01-23 The Broad Institute, Inc. Compounds, Compositions, and Methods for Cancer Therapy
US9108923B2 (en) * 2012-07-20 2015-08-18 Howard Hughes Medical Institute Compounds, compositions, and methods for cancer therapy
US10766865B2 (en) 2012-10-16 2020-09-08 Sumitomo Dainippon Pharma Oncology, Inc. PKM2 modulators and methods for their use
US11712433B2 (en) 2019-03-22 2023-08-01 Sumitomo Pharma Oncology, Inc. Compositions comprising PKM2 modulators and methods of treatment using the same

Also Published As

Publication number Publication date
WO2012030408A1 (en) 2012-03-08
US20110053938A1 (en) 2011-03-03

Similar Documents

Publication Publication Date Title
US8349832B2 (en) Compounds and compositions for treating cancer
US8318737B2 (en) Compounds and compositions for treating cancer
JP5421340B2 (en) Use of Asian acid or Asian chicoside for cancer treatment.
JP4172726B2 (en) Formulation containing a covalent complex of cis-docosahexaenoic acid and docetaxel
US5919815A (en) Taxane compounds and compositions
US20080125380A1 (en) Fatty acid-anticancer conjugates and uses thereof
CA2667684C (en) Hypoxia-selective, weakly basic 2-nitroimidazole delivery agents and methods of use thereof
US20090312373A1 (en) Methods for the treatment of cancer using piperlongumine and piperlongumine analogs
US20100129363A1 (en) Methods and compositions using pde4 inhibitors for the treatment and management of cancers
US10604510B2 (en) Solid forms comprising (1E, 4E)-2-amino-N,N-dipropyl-8-(4-(pyrrolidine-1-carbonyl)phenyl)-3H-benzo[B]azepine-4-carboxami compositions thereof, and uses thereof
AU2013345007A1 (en) Gemcitabine prodrugs and uses thereof
EP2473506A2 (en) Compounds and compositions for treating cancer
US20130237539A1 (en) Compounds and Compositions for Treating Cancer
WO2000067802A1 (en) Fatty acid-n-substituted indol-3-glyoxyl-amide compositions and uses thereof
CA2677813A1 (en) Method of treating cell proliferative disorders using growth hormone secretagogues
US20060199835A1 (en) Neuroprotective and anti-proliferative compounds
US10646479B2 (en) Use of N-(4-iodobenzoylamino)-5-ethyl-1,2,3,6-tetrahydropyridine as a treatment for cancer
WO2006063762A1 (en) Dipeptide nitriles

Legal Events

Date Code Title Description
AS Assignment

Owner name: CANTHERA THERAPEUTICS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FOLEY, MICHAEL A;GOULD, ROBERT;ELLIOTT, PETER;AND OTHERS;SIGNING DATES FROM 20100329 TO 20100430;REEL/FRAME:027537/0594

AS Assignment

Owner name: MASSACHUSETTS GENERAL HOSPITAL CORPORATION, MASSAC

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, SAM;REEL/FRAME:027769/0832

Effective date: 20120105

Owner name: MASSACHUSETTS GENERAL HOSPITAL CORPORATION, MASSAC

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MANDINOVA, ANNA;REEL/FRAME:027769/0776

Effective date: 20120105

Owner name: LEE, SAM, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CANTHERA THERAPEUTICS INC.;REEL/FRAME:027769/0740

Effective date: 20111220

Owner name: MANDINOVA, ANNA, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CANTHERA THERAPEUTICS INC.;REEL/FRAME:027769/0680

Effective date: 20111220

AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LEE, SAM W.;REEL/FRAME:028448/0948

Effective date: 20120626

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MANDINOVA, ANNA;REEL/FRAME:028448/0983

Effective date: 20120626

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CANTHERA THERAPEUTICS, INC.;ONCOBIOSCIENCES CO. LTD.;REEL/FRAME:032473/0798

Effective date: 20131220

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2552); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 8